DOI QR코드

DOI QR Code

Intrathecal administration of naringenin improves motor dysfunction and neuropathic pain following compression spinal cord injury in rats: relevance to its antioxidant and anti-inflammatory activities

  • Fakhri, Sajad (Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences) ;
  • Sabouri, Shahryar (Student Research Committee, Kermanshah University of Medical Sciences) ;
  • Kiani, Amir (Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences) ;
  • Farzaei, Mohammad Hosein (Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences) ;
  • Rashidi, Khodabakhsh (Research Center of Oils and Fats, Kermanshah University of Medical Sciences) ;
  • Mohammadi-Farani, Ahmad (Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences) ;
  • Mohammadi-Noori, Ehsan (Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences) ;
  • Abbaszadeh, Fatemeh (Department of Neuroscience, Faculty of Advanced Technologies in Medical Sciences, Iran University of Medical Sciences)
  • Received : 2022.03.28
  • Accepted : 2022.05.13
  • Published : 2022.07.01

Abstract

Background: Spinal cord injury (SCI) is one of the most debilitating disorders throughout the world, causing persistent sensory-motor dysfunction, with no effective treatment. Oxidative stress and inflammatory responses play key roles in the secondary phase of SCI. Naringenin (NAR) is a natural flavonoid with known anti-inflammatory and antioxidative properties. This study aims at evaluating the effects of intrathecal NAR administration on sensory-motor disability after SCI. Methods: Animals underwent a severe compression injury using an aneurysm clip. About 30 minutes after surgery, NAR was injected intrathecally at the doses of 5, 10, and 15 mM in 20 µL volumes. For the assessment of neuropathic pain and locomotor function, acetone drop, hot plate, inclined plane, and Basso, Beattie, Bresnahan tests were carried out weekly till day 28 post-SCI. Effects of NAR on matrix metalloproteinase (MMP)-2 and MMP-9 activity was appraised by gelatin zymography. Also, histopathological analyses and serum levels of glutathione (GSH), catalase and nitrite were measured in different groups. Results: NAR reduced neuropathic pain, improved locomotor function, and also attenuated SCI-induced weight loss weekly till day 28 post-SCI. Zymography analysis showed that NAR suppressed MMP-9 activity, whereas it increased that of MMP-2, indicating its anti-neuroinflammatory effects. Also, intrathecal NAR modified oxidative stress related markers GSH, catalase, and nitrite levels. Besides, the neuroprotective effect of NAR was corroborated through increased survival of sensory and motor neurons after SCI. Conclusions: These results suggest intrathecal NAR as a promising candidate for medical therapeutics for SCI-induced sensory and motor dysfunction.

Keywords

Acknowledgement

The authors acknowledged Pharmaceutical Sciences Research Center, Kermanshah University of Medical Sciences, for the special Grant No. 4000170.

References

  1. Thuret S, Moon LD, Gage FH. Therapeutic interventions after spinal cord injury. Nat Rev Neurosci 2006; 7: 628-43. Erratum in: Nat Rev Neurosci 2006; 7: 902. https://doi.org/10.1038/nrn1955
  2. Fakhri S, Dargahi L, Abbaszadeh F, Jorjani M. Effects of astaxanthin on sensory-motor function in a compression model of spinal cord injury: Involvement of ERK and AKT signalling pathway. Eur J Pain 2019; 23: 750-64. https://doi.org/10.1002/ejp.1342
  3. Fakhri S, Dargahi L, Abbaszadeh F, Jorjani M. Astaxanthin attenuates neuroinflammation contributed to the neuropathic pain and motor dysfunction following compression spinal cord injury. Brain Res Bull 2018; 143: 217-24. https://doi.org/10.1016/j.brainresbull.2018.09.011
  4. Naseem M, Parvez S. Role of melatonin in traumatic brain injury and spinal cord injury. ScientificWorldJournal 2014; 2014: 586270.
  5. Esposito E, Genovese T, Caminiti R, Bramanti P, Meli R, Cuzzocrea S. Melatonin regulates matrix metalloproteinases after traumatic experimental spinal cord injury. J Pineal Res 2008; 45: 149-56. https://doi.org/10.1111/j.1600-079X.2008.00569.x
  6. Jia Z, Zhu H, Li J, Wang X, Misra H, Li Y. Oxidative stress in spinal cord injury and antioxidant-based intervention. Spinal Cord 2012; 50: 264-74. https://doi.org/10.1038/sc.2011.111
  7. Faden AI, Wu J, Stoica BA, Loane DJ. Progressive inflammation-mediated neurodegeneration after traumatic brain or spinal cord injury. Br J Pharmacol 2016; 173: 681-91. https://doi.org/10.1111/bph.13179
  8. Franz S, Finnerup NB. Diagnostics and treatment of pain in spinal cord injury. In: Neurological aspects of spinal cord injury. Edited by Weidner N, Rupp R, Tansey K. Cham, Springer. 2017, pp 283-302.
  9. Backonja MM, Irving G, Argoff C. Rational multidrug therapy in the treatment of neuropathic pain. Curr Pain Headache Rep 2006; 10: 34-8. https://doi.org/10.1007/s11916-006-0007-1
  10. Hare JT, Elliott DP. Grapefruit juice and potential drug interactions. Consult Pharm 2003; 18: 466-72.
  11. Palma-Duran SA, Caire-Juvera G, Robles-Burgeno Mdel R, Ortega-Velez MI, Gutierrez-Coronado Mde L, Almada Mdel C, et al. Serum levels of phytoestrogens as biomarkers of intake in Mexican women. Int J Food Sci Nutr 2015; 66: 819-25. https://doi.org/10.3109/09637486.2015.1092019
  12. Pinho-Ribeiro FA, Zarpelon AC, Fattori V, Manchope MF, Mizokami SS, Casagrande R, et al. Naringenin reduces inflammatory pain in mice. Neuropharmacology 2016; 105: 508-19. https://doi.org/10.1016/j.neuropharm.2016.02.019
  13. Hu CY, Zhao YT. Analgesic effects of naringenin in rats with spinal nerve ligation-induced neuropathic pain. Biomed Rep 2014; 2: 569-73. https://doi.org/10.3892/br.2014.267
  14. Al-Rejaie SS, Aleisa AM, Abuohashish HM, Parmar MY, Ola MS, Al-Hosaini AA, et al. Naringenin neutralises oxidative stress and nerve growth factor discrepancy in experimental diabetic neuropathy. Neurol Res 2015; 37: 924-33. https://doi.org/10.1179/1743132815Y.0000000079
  15. Kaulaskar S, Bhutada P, Rahigude A, Jain D, Harle U. Effects of naringenin on allodynia and hyperalgesia in rats with chronic constriction injury-induced neuropathic pain. Zhong Xi Yi Jie He Xue Bao 2012; 10: 1482-9. https://doi.org/10.3736/jcim20121223
  16. Pinho-Ribeiro FA, Zarpelon AC, Mizokami SS, Borghi SM, Bordignon J, Silva RL, et al. The citrus flavonone naringenin reduces lipopolysaccharide-induced inflammatory pain and leukocyte recruitment by inhibiting NF-κB activation. J Nutr Biochem 2016; 33: 8-14. https://doi.org/10.1016/j.jnutbio.2016.03.013
  17. Manchope MF, Calixto-Campos C, Coelho-Silva L, Zarpelon AC, Pinho-Ribeiro FA, Georgetti SR, et al. Naringenin inhibits superoxide anion-induced inflammatory pain: role of oxidative stress, cytokines, Nrf-2 and the NO-cGMP-PKG-KATP channel signaling pathway. PLoS One 2016; 11: e0153015. https://doi.org/10.1371/journal.pone.0153015
  18. Fakhri S, Kiani A, Jalili C, Abbaszadeh F, Piri S, Farzaei MH, et al. Intrathecal administration of melatonin ameliorates the neuroinflammation- mediated sensory and motor dysfunction in a rat model of compression spinal cord injury. Curr Mol Pharmacol 2021; 14: 646-57. https://doi.org/10.2174/1874467213666201230101811
  19. Mestre C, Pelissier T, Fialip J, Wilcox G, Eschalier A. A method to perform direct transcutaneous intrathecal injection in rats. J Pharmacol Toxicol Methods 1994; 32: 197-200. https://doi.org/10.1016/1056-8719(94)90087-6
  20. Kauppila T. Cold exposure enhances tactile allodynia transiently in mononeuropathic rats. Exp Neurol 2000; 161: 740-4. https://doi.org/10.1006/exnr.1999.7287
  21. Hama AT, Plum AW, Sagen J. Antinociceptive effect of ambroxol in rats with neuropathic spinal cord injury pain. Pharmacol Biochem Behav 2010; 97: 249-55. https://doi.org/10.1016/j.pbb.2010.08.006
  22. Janicki P, Libich J. Detection of antagonist activity for narcotic analgesics in mouse hot-plate test. Pharmacol Biochem Behav 1979; 10: 623-6. https://doi.org/10.1016/0091-3057(79)90244-2
  23. Basso DM, Beattie MS, Bresnahan JC. A sensitive and reliable locomotor rating scale for open field testing in rats. J Neurotrauma 1995; 12: 1-21. https://doi.org/10.1089/neu.1995.12.1
  24. Samini F, Samarghandian S, Borji A, Mohammadi G, bakaian M. Curcumin pretreatment attenuates brain lesion size and improves neurological function following traumatic brain injury in the rat. Pharmacol Biochem Behav 2013; 110: 238-44. https://doi.org/10.1016/j.pbb.2013.07.019
  25. Rahman I, Kode A, Biswas SK. Assay for quantitative determination of glutathione and glutathione disulfide levels using enzymatic recycling method. Nat Protoc 2006; 1: 3159-65. https://doi.org/10.1038/nprot.2006.378
  26. Aebi H. Catalase. In: Methods of enzymatic analysis. 2nd ed. Edited by Bergmeyer HU. New York, Academic Press. 1974, pp 673-84.
  27. Masoudi A, Dargahi L, Abbaszadeh F, Pourgholami MH, Asgari A, Manoochehri M, et al. Neuroprotective effects of astaxanthin in a rat model of spinal cord injury. Behav Brain Res 2017; 329: 104-10. https://doi.org/10.1016/j.bbr.2017.04.026
  28. Nouri Z, Fakhri S, El-Senduny FF, Sanadgol N, Abd-ElGhani GE, Farzaei MH, et al. On the neuroprotective effects of naringenin: pharmacological targets, signaling pathways, molecular mechanisms, and clinical perspective. Biomolecules 2019; 9: 690. https://doi.org/10.3390/biom9110690
  29. Mao L, Wang H, Qiao L, Wang X. Disruption of Nrf2 enhances the upregulation of nuclear factor-kappaB activity, tumor necrosis factor-α, and matrix metalloproteinase-9 after spinal cord injury in mice. Mediators Inflamm 2010; 2010: 238321. https://doi.org/10.1155/2010/238321
  30. Alghamdi BS. The neuroprotective role of melatonin in neurological disorders. J Neurosci Res 2018; 96: 1136-49. https://doi.org/10.1002/jnr.24220
  31. Stetler-Stevenson WG. Dynamics of matrix turnover during pathologic remodeling of the extracellular matrix. Am J Pathol 1996; 148: 1345-50.
  32. Shibata N, Ohnuma T, Higashi S, Usui C, Ohkubo T, Kitajima A, et al. Genetic association between matrix metalloproteinase MMP-9 and MMP-3 polymorphisms and Japanese sporadic Alzheimer's disease. Neurobiol Aging 2005; 26: 1011-4. https://doi.org/10.1016/j.neurobiolaging.2004.09.004
  33. Lorenzl S, Albers DS, Narr S, Chirichigno J, Beal MF. Expression of MMP-2, MMP-9, and MMP-1 and their endogenous counterregulators TIMP-1 and TIMP-2 in postmortem brain tissue of Parkinson's disease. Exp Neurol 2002; 178: 13-20. https://doi.org/10.1006/exnr.2002.8019
  34. Beuche W, Yushchenko M, Mader M, Maliszewska M, Felgenhauer K, Weber F. Matrix metalloproteinase-9 is elevated in serum of patients with amyotrophic lateral sclerosis. Neuroreport 2000; 11: 3419-22. https://doi.org/10.1097/00001756-200011090-00003
  35. Noble LJ, Donovan F, Igarashi T, Goussev S, Werb Z. Matrix metalloproteinases limit functional recovery after spinal cord injury by modulation of early vascular events. J Neurosci 2002; 22: 7526-35. https://doi.org/10.1523/jneurosci.22-17-07526.2002
  36. Yang R, Guo L, Wang P, Huang L, Tang Y, Wang W, et al. Epidemiology of spinal cord injuries and risk factors for complete injuries in Guangdong, China: a retrospective study. PLoS One 2014; 9: e84733. https://doi.org/10.1371/journal.pone.0084733
  37. Wells JE, Rice TK, Nuttall RK, Edwards DR, Zekki H, Rivest S, et al. An adverse role for matrix metalloproteinase 12 after spinal cord injury in mice. J Neurosci 2003; 23: 10107-15. https://doi.org/10.1523/jneurosci.23-31-10107.2003
  38. Zhang H, Chang M, Hansen CN, Basso DM, Noble-Haeusslein LJ. Role of matrix metalloproteinases and therapeutic benefits of their inhibition in spinal cord injury. Neurotherapeutics 2011; 8: 206-20. https://doi.org/10.1007/s13311-011-0038-0
  39. Piao MS, Lee JK, Jang JW, Hur H, Lee SS, Xiao L, et al. Melatonin improves functional outcome via inhibition of matrix metalloproteinases-9 after photothrombotic spinal cord injury in rats. Acta Neurochir (Wien) 2014; 156: 2173-82. https://doi.org/10.1007/s00701-014-2119-4
  40. Jang JW, Lee JK, Kim SH. Activation of matrix metalloproteinases-9 after photothrombotic spinal cord injury model in rats. J Korean Neurosurg Soc 2011; 50: 288-92. https://doi.org/10.3340/jkns.2011.50.4.288
  41. Bai X, Zhang X, Chen L, Zhang J, Zhang L, Zhao X, et al. Protective effect of naringenin in experimental ischemic stroke: down-regulated NOD2, RIP2, NF-κB, MMP-9 and up-regulated claudin-5 expression. Neurochem Res 2014; 39: 1405-15. https://doi.org/10.1007/s11064-014-1326-y
  42. Hsu JY, McKeon R, Goussev S, Werb Z, Lee JU, Trivedi A, et al. Matrix metalloproteinase-2 facilitates wound healing events that promote functional recovery after spinal cord injury. J Neurosci 2006; 26: 9841-50. https://doi.org/10.1523/jneurosci.1993-06.2006
  43. Shi LB, Tang PF, Zhang W, Zhao YP, Zhang LC, Zhang H. Naringenin inhibits spinal cord injury-induced activation of neutrophils through miR-223. Gene 2016; 592: 128-33. https://doi.org/10.1016/j.gene.2016.07.037
  44. Puspitasari V, Gunawan PY, Wiradarma HD, Hartoyo V. Glial fibrillary acidic protein serum level as a predictor of clinical outcome in ischemic stroke. Open Access Maced J Med Sci 2019; 7: 1471-4. https://doi.org/10.3889/oamjms.2019.326
  45. Khajevand-Khazaei MR, Ziaee P, Motevalizadeh SA, Rohani M, Afshin-Majd S, Baluchnejadmojarad T, et al. Naringenin ameliorates learning and memory impairment following systemic lipopolysaccharide challenge in the rat. Eur J Pharmacol 2018; 826: 114-22. https://doi.org/10.1016/j.ejphar.2018.03.001
  46. Krishna Chandran AM, Christina H, Das S, Mumbrekar KD, Satish Rao BS. Neuroprotective role of naringenin against methylmercury induced cognitive impairment and mitochondrial damage in a mouse model. Environ Toxicol Pharmacol 2019; 71: 103224. https://doi.org/10.1016/j.etap.2019.103224
  47. Chtourou Y, Fetoui H, Gdoura R. Protective effects of naringenin on iron-overload-induced cerebral cortex neurotoxicity correlated with oxidative stress. Biol Trace Elem Res 2014; 158: 376-83. https://doi.org/10.1007/s12011-014-9948-0
  48. Muthaiah VP, Venkitasamy L, Michael FM, Chandrasekar K, Venkatachalam S. Neuroprotective role of naringenin on carbaryl induced neurotoxicity in mouse neuroblastoma cells. J Pharmacol Pharmacother 2013; 4: 192-7. https://doi.org/10.4103/0976-500X.114599
  49. Xu XH, Ma CM, Han YZ, Li Y, Liu C, Duan ZH, et al. Protective effect of naringenin on glutamate-induced neurotoxicity in cultured hippocampal cells. Arch Biol Sci 2015; 67: 639-46. https://doi.org/10.2298/ABS140811023X
  50. Giangregorio L, McCartney N. Bone loss and muscle atrophy in spinal cord injury: epidemiology, fracture prediction, and rehabilitation strategies. J Spinal Cord Med 2006; 29: 489-500. https://doi.org/10.1080/10790268.2006.11753898